Mitochondria Transcription Factor A: A Putative Target for the Effect of Melatonin on U87MG Malignant Glioma Cell Line - MDPI

Page created by Brad Edwards
 
CONTINUE READING
Mitochondria Transcription Factor A: A Putative Target for the Effect of Melatonin on U87MG Malignant Glioma Cell Line - MDPI
molecules
Article
Mitochondria Transcription Factor A: A Putative
Target for the Effect of Melatonin on U87MG
Malignant Glioma Cell Line
Daiane G. Franco *      ID
                             , Isabele F. Moretti and Suely K. N. Marie
 Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP 01246903, Brazil;
 isabelemoretti@gmail.com (I.F.M.); sknmarie@usp.br (S.K.N.M.)
 * Correspondence: daianegfranco@yahoo.com.br; Tel.: +55-11-30618559

 Academic Editor: Dun-Xian Tan
                                                                                                 
 Received: 20 February 2018; Accepted: 7 May 2018; Published: 9 May 2018                         

 Abstract: The disruption of mitochondrial activity has been associated with cancer development
 because it contributes to regulating apoptosis and is the main source of reactive oxygen species (ROS)
 production. Mitochondrial transcription factor A (TFAM) is a protein that maintains mitochondrial
 DNA (mtDNA) integrity, and alterations in its expression are associated with mitochondrial damage
 and cancer development. In addition, studies have shown that mitochondria are a known target
 of melatonin, the pineal gland hormone that plays an important anti-tumorigenic role. Thus,
 we hypothesized that melatonin decreases the expression of TFAM (RNA and protein) in the human
 glioblastoma cell line U87MG, which disrupts mtDNA expression and results in cell death due to
 increased ROS production and mitochondrial damage. Our results confirm the hypothesis, and also
 show that melatonin reduced the expression of other mitochondrial transcription factors mRNA
 (TFB1M and TFB2M) and interfered with mtDNA transcription. Moreover, melatonin delayed
 cell cycle progression and potentiated the reduction of cell survival due to treatment with the
 chemotherapeutic agent temozolomide. In conclusion, elucidating the effect of melatonin on TFAM
 expression should help to understand the signaling pathways involved in glioblastoma progression,
 and melatonin could be potentially applied in the treatment of this type of brain tumor.

 Keywords: melatonin; mitochondria; TFAM; cancer; glioblastoma

1. Introduction
     Mitochondria are double-membrane organelles that contain their own genetic material,
mitochondrial DNA (mtDNA), which is circular, high-copy-number DNA [1,2]. In addition to
being an energy source for the cell that produces adenosine triphosphate (ATP) via oxidative
phosphorylation, mitochondria are important in other processes, such as calcium homeostasis,
fatty acid oxidation, the synthesis of reactive oxygen species (ROS), apoptosis, the cell cycle, and
proliferation. Accordingly, the disruption of mitochondrial activity is associated with several diseases,
including cancer [3–5]. Moreover, mtDNA associates with the internal membrane of mitochondria
via a nucleoprotein complex called nucleoid [6,7], and the most abundant protein component of this
structure is mitochondrial transcription factor A (TFAM), which belongs to the high mobility group
(HMG) protein family [7–9]. Although TFAM can bind to mtDNA in a nonspecific manner to promote
the packaging and maintenance of genetic material, TFAM also binds to the promoter region to regulate
transcription, together with two other transcription factors (TFAB1M and TFB2M) that form a complex
with mitochondrial RNA polymerase (POLRMT) [10]. In addition, TFAM also regulates the replication
of mtDNA by controlling its copy number [6,11].

Molecules 2018, 23, 1129; doi:10.3390/molecules23051129                    www.mdpi.com/journal/molecules
Molecules 2018, 23, 1129                                                                             2 of 16

      TFAM has been considered a potential target for cancer therapy since changes in its expression
have been detected in several types of cancer [12–15]. In glioma, the TFAM RNA and protein levels
are upregulated, compared to non-neoplastic brain tissue [12,13]. Nevertheless, the protein levels of
TFAM positively correlated with the malignancy of glioma [15], higher RNA levels of TFAM correlated
with a better prognosis among patients with grade IV glioma (glioblastoma, GBM) [13].
      Glioblastoma, or grade IV astrocytoma, as classified by the World Health Organization (WHO),
is a tumor of glial origin restricted to the central nervous system (CNS) that is highly invasive
to the surrounding cerebral parenchyma [16,17]. Despite advances in knowledge regarding the
molecular biology of astrocytomas that have improved diagnosis and treatment, the prognosis of this
condition remains poor, with a maximum survival of 24 months [16–18]. The treatment of this cancer
currently involves a combination of surgery, radiotherapy, and chemotherapy with temozolomide
(TMZ) [16,19–22].
      Melatonin (N-acetyl-5-methoxytryptamine), a hormone synthesized from serotonin, acts on
several cellular processes, including proliferation, differentiation, invasion, and apoptosis, to result in
different effects on non-tumor cells and tumor cells [23,24]. In normal cells, melatonin increases viability
and acts as an antioxidant, whereas it activates apoptosis and increases the cellular content of reactive
oxygen species (ROS) in tumor cells, effects that depend on mitochondrial activity [25,26]. The use
of melatonin as an adjuvant cancer chemotherapy has shown promising results in relation to both
improving the efficacy of treatment and reducing the incidence of side effects. The effects of melatonin
on mitochondria have been widely explored, however little is known about the effects of melatonin
on mtDNA and TFAM expression. We hypothesized that melatonin can reduce the expression of
mitochondrial transcription factors (TFAM, TFMB1M, and TFB2M) to decrease the viability of cancer
cells due to an imbalance in mitochondrial activity. Using a GBM cell line (U87MG), we showed that
melatonin decreased the level of mitochondrial transcription factors, induced mitochondrial membrane
depolarization to cause apoptosis, increased intracellular oxidative stress, and delayed the cell cycle.
When used as an adjuvant therapy with TMZ, melatonin enhanced the efficacy of chemotherapy by
further decreasing cell viability/proliferation.

2. Results

2.1. Melatonin Decreased the Expression of TFAM, TFB1M, and TFB2M
     We first investigated the ability of melatonin to change the gene expression of transcription factors
that act on mitochondria, and the effect of this change on the proper functioning of the organelle and,
consequently, the cell. Incubation with melatonin (1 mM or 3 mM) for 72 h reduced the expression of the
transcriptions factors TFAM (Vehicle: 1.01 ± 0.05%; Mel 1 mM: 0.73 ± 0.10%; Mel 3 mM: 0.66 ± 0.07%),
TFB1M (Vehicle: 1.04 ± 0.06%; Mel 1 mM: 0.46 ± 0.05%; Mel 3 mM: 0.41 ± 0.07%), and TFB2M (Vehicle:
1.02 ± 0.05%; Mel 1 mM: 0.50 ± 0.03%; Mel 3 mM: 0.47 ± 0.10%), compared to the vehicle control
group (Figure 1A–C).

2.2. Melatonin Decreased the Content of TFAM Protein
     Western blotting analysis showed that expression of TFAM at the protein level was decreased
following melatonin (3 mM) treatment compared to the vehicle group (ethanol 0.9%), but for the
1 mM concentration, the melatonin effect was variable and the result was not statistically significant.
(Figure 2 and Supplementary material—Figure S1, Table S2).
organelle and, consequently, the cell. Incubation with melatonin (1 mM or 3 mM) for 72 h reduced
the expression of the transcriptions factors TFAM (Vehicle: 1.01 ± 0.05%; Mel 1 mM: 0.73 ± 0.10%; Mel
3 mM: 0.66 ± 0.07%), TFB1M (Vehicle: 1.04 ± 0.06%; Mel 1 mM: 0.46 ± 0.05%; Mel 3 mM: 0.41 ± 0.07%),
and TFB2M (Vehicle: 1.02 ± 0.05%; Mel 1 mM: 0.50 ± 0.03%; Mel 3 mM: 0.47 ± 0.10%), compared to the
   Molecules
vehicle      2018, 23,
         control       1129 (Figure 1A–C).
                    group                                                                  3 of 16

  Molecules 2018, 23, x FOR PEER REVIEW                                                                                3 of 16

       Figure 1. Melatonin inhibits the expression of mitochondrial transcription factor A (TFAM), TFB1M,
       and TFB2M—Cultured U87MG cells were incubated with melatonin (1 mM or 3 mM) for 72 h, and
       the medium was exchanged every 24 h. The relative mRNA expression levels of each gene were
       quantified by qRT-PCR using the geometric mean of the following normalizing genes: Hypoxanthime
       phosphoribosyl transferase (HPRT), glucuronidase-beta (GUS-B), and TATA-Box binding protein
       (TBP) [27]. The data are expressed as the relative quantification (2−ΔΔCt) compared to the vehicle-
       treated groups (ethanol 0.3% or 0.9%). Gene expression did not differ in cells treated with vehicle or
        Figure
       0.3%  and   Melatonin
                1. 0.9%        inhibits
                         ethanol,        the expression
                                   and these   groups wereof mitochondrial
                                                              represented astranscription
                                                                                a single group.factor A (TFAM),
                                                                                                   From            TFB1M,
                                                                                                         left to right are
        and TFB2M—Cultured
       presented    the results for U87MG
                                    TFAM (A),cells were incubated
                                                 TFB1M               with melatonin
                                                          (B), and TFB2M                 (1 mM
                                                                             (C). * p < 0.05,     or 3with
                                                                                              tested  mM)anfor   72 h, and
                                                                                                               analysis of
        the medium     was   exchanged    every   24 h.  The  relative mRNA      expression    levels of
       variance followed by the Bonferroni post-hoc correction using GraphPad Prism version 5, comparing
                                                                                                  ®      each   gene were
        quantified
       the effect ofby  qRT-PCRtousing
                      melatonin           the geometric
                                     the vehicle  group. mean of the following normalizing genes: Hypoxanthime
        phosphoribosyl transferase (HPRT), glucuronidase-beta (GUS-B), and TATA-Box binding protein
        (TBP) [27].Decreased
  2.2. Melatonin     The data are
                                theexpressed
                                    Content ofas TFAM
                                                 the relative quantification (2−∆∆Ct ) compared to the vehicle-treated
                                                          Protein
        groups (ethanol 0.3% or 0.9%). Gene expression did not differ in cells treated with vehicle or 0.3% and
       Western blotting analysis showed that expression of TFAM at the protein level was decreased
        0.9% ethanol, and these groups were represented as a single group. From left to right are presented
  following   melatonin
        the results         (3 mM)
                     for TFAM          treatment
                                 (A), TFB1M          compared
                                               (B), and           to the
                                                         TFB2M (C).    * p  0.05 compared to vehicle. The statistical analysis consisted of an ANOVA followed by Bonferroni’s
        represented as a single bar. * p > 0.05 compared to vehicle. The statistical analysis consisted of an
         post-hoc test.
        ANOVA followed by Bonferroni’s post-hoc test.

  2.3. Melatonin Decreased the Transcription of mtDNA but Did Not Affect Replication
  2.3. Melatonin Decreased the Transcription of mtDNA but Did Not Affect Replication
        Since the transcription factors TFAM, TFB1M, and TFB2M are directly related to the regulation of
       Since the transcription factors TFAM, TFB1M, and TFB2M are directly related to the regulation
  transcription and mtDNA replication, we evaluated the expression of the MT-ND1 gene to ascertain
  of transcription and mtDNA replication, we evaluated the expression of the MT-ND1 gene to
  if the effect of melatonin on transcription factors was reflected in mitochondrial gene expression
  ascertain if the effect of melatonin on transcription factors was reflected in mitochondrial gene
  and mtDNA copy number. To examine mitochondrial gene expression and mtDNA copy number,
  expression and mtDNA copy number. To examine mitochondrial gene expression and mtDNA copy
  we used the aforementioned primer for the NADH dehydrogenase 1 gene and mRNA and DNA
  number, we used the aforementioned primer for the NADH dehydrogenase 1 gene and mRNA and
  extracted from U87MG cells treated with 1 mM or 3 mM of melatonin for 72 h, respectively. Melatonin
  DNA extracted from U87MG cells treated with 1 mM or 3 mM of melatonin for 72 h, respectively.
  reduced the expression of the mtDNA gene MT-ND1 (Vehicle: 1.01 ± 0.05%; Mel 1 mM: 0.54 ± 0.06%;
  Melatonin reduced the expression of the mtDNA gene MT-ND1 (Vehicle: 1.01 ± 0.05%; Mel 1 mM:
  Mel 3 mM: 0.62 ± 0.12%) (Figure 3A), but despite the reduction in TFAM, TFB1M, and TFB2M
  0.54 ± 0.06%; Mel 3 mM: 0.62 ± 0.12%) (Figure 3A), but despite the reduction in TFAM, TFB1M, and
  TFB2M expression, mtDNA replication appeared unchanged, since the number of copies of
  mitochondrial genetic material remained the same after treatment with melatonin (Figure 3B).
Molecules 2018, 23, 1129                                                                                               4 of 16

expression, mtDNA replication appeared unchanged, since the number of copies of mitochondrial
  Molecules 2018, 23, x FOR PEER REVIEW                                                  4 of 16
genetic material remained the same after treatment with melatonin (Figure 3B).

      Figure 3. Melatonin inhibits mitochondrial NADH dehydrogenase 1 gene expression but does not affect
         Figure 3. Melatonin inhibits mitochondrial NADH dehydrogenase 1 gene expression but does not
      mitochondrial DNA (mtDNA) replication—Cultured U87MG cells were incubated with melatonin
         affect mitochondrial DNA (mtDNA) replication—Cultured U87MG cells were incubated with
      (1 mM or 3 mM) for 72 h, and the medium was exchanged every 24 h. The relative expression of
         melatonin (1 mM or 3 mM) for 72 h, and the medium was exchanged every 24 h. The relative
      the NADH dehydrogenase 1 gene (A) and mtDNA copy number (B) were determined by qRT-PCR,
         expression
      using           of the NADH
              mitochondrial    RNA and  dehydrogenase      1 gene respectively.
                                          DNA as a template,      (A) and mtDNA      copyare
                                                                                 The data  number    (B) were
                                                                                             expressed         determined
                                                                                                          as the relative
         by  qRT-PCR,
      quantification (2  using
                         − ∆∆Ct  mitochondrial   RNA     and  DNA   as a template,  respectively. The  data  are
                                ) compared to the vehicle-treated groups (ethanol 0.3% or 0.9%). Gene expression expressed
      didasnot
            thediffer
                relative  quantification
                               treated with(2vehicle
                                              −ΔΔCt) compared to the vehicle-treated groups (ethanol 0.3% or 0.9%).
                      in cells                         or 0.3% and 0.9% ethanol, and these groups were represented
      as Gene  expression
          a single bar * p < did  not tested
                                0.05, differ in cellsan
                                              with    treated  with
                                                        analysis  ofvehicle or 0.3%
                                                                     variance        and 0.9%
                                                                                followed       ethanol,
                                                                                          by the         and these
                                                                                                  Bonferroni        groups
                                                                                                               post-hoc
         were represented
      correction               as a single
                  using GraphPad       Prismbar   * p < 5,
                                              ® version   0.05, tested with
                                                             comparing        an analysis
                                                                         the effect        of variance
                                                                                    of melatonin         followed
                                                                                                  to the vehicle     by the
                                                                                                                  group.
         Bonferroni post-hoc correction using GraphPad Prism® version 5, comparing the effect of melatonin
         to the vehicle group.
2.4. Melatonin Induced ROS Production

  2.4.ToMelatonin
         verify that melatonin
                  Induced ROSincreases    oxidative stress in U87MG cells, we evaluated the production of
                                 Production
superoxides as an indicator of ROS production using cytometry, based on the reaction of total cellular
        To verify that melatonin increases oxidative stress in U87MG cells, we evaluated the production
superoxide with dihydroethidium (DHE). The result showed that melatonin increased ROS production
  of superoxides as an indicator of ROS production using cytometry, based on the reaction of total
to 20.73 ± 1.03% at a concentration of 1 mM, and 23.62 ± 4.56% at a concentration of 3 mM, compared
  cellular superoxide with dihydroethidium (DHE). The result showed that melatonin increased ROS
to the vehicle group (14.97 ± 1.89%, Figure 4A). To verify if the increase of ROS induced by melatonin
  production to 20.73 ± 1.03% at a concentration of 1 mM, and 23.62 ± 4.56% at a concentration of 3 mM,
has an effect on cell viability, a known ROS scavenger, N-acetyl-L-cysteine (NAC, 10 mM), was used.
  compared to the vehicle group (14.97 ± 1.89%, Figure 4A). To verify if the increase of ROS induced by
Figure 4B shows that the antioxidant agent reverts the 1 mM melatonin-induced viability reduction
  melatonin has an effect on cell viability, a known ROS scavenger, N-acetyl-L-cysteine (NAC, 10 mM),
and about 40% the effect of melatonin 3 mM.
  was used. Figure 4B shows that the antioxidant agent reverts the 1 mM melatonin-induced viability
  reduction
2.5. Melatoninand  aboutMitochondria
                Induced  40% the effect  of melatoninand
                                      Depolarization    3 mM.
                                                          Apoptosis
      Two different cytometry experiments were performed to assess the ability of melatonin to induce
cell death. First, the depolarization of mitochondria was measured to verify if the loss of mitochondrial
inner membrane potential is associated with the early stage of apoptosis. The incubation of U87MG
with 1 mM and 3 mM melatonin for 72 h decreased the percentage of live cells in the vehicle group
from 51.55 ± 047% to 41.25 ± 3.61% and 14.14 ± 9.00%, respectively. Moreover, the percentage of cells
with depolarized mitochondria increased from 44.25 ± 0.4% in the vehicle group to 53.65 ± 4.60% and
78.66 ± 13.87% in the melatonin-treated groups (Figure 5A).
      Second, apoptotic and necrotic cells were measured by staining phosphatidyl serine with Annexin
V and 7-AAD, respectively. The results showed that the percentage of live cells in the vehicle group
86.99 ± 1.42% decreased to 83.31 ± 3.80% and 78.63 ± 3.71% in the groups treated with 1 mM and 3 mM
melatonin, respectively. Specifically, the number of apoptotic cells, but not necrotic cells, increased.
Apoptotic cells increased to 11.79 ± 3.61% and 16.27 ± 4.86% in the melatonin group, whereas the
proportion of apoptotic cells was 8.26 ± 1.71% in the vehicle group (Figure 5B). The rates of apoptotic

        Figure 4. Melatonin increases reactive oxygen species (ROS) production—(A) U87MG cells were
        incubated with melatonin (1 mM or 3 mM) for 72 h, and the medium was exchanged every 24 h. ROS
To verify that melatonin increases oxidative stress in U87MG cells, we evaluated the production
of superoxides as an indicator of ROS production using cytometry, based on the reaction of total
cellular superoxide with dihydroethidium (DHE). The result showed that melatonin increased ROS
production to 20.73 ± 1.03% at a concentration of 1 mM, and 23.62 ± 4.56% at a concentration of 3 mM,
 Molecules 2018, 23, 1129                                                                               5 of 16
compared to the vehicle group (14.97 ± 1.89%, Figure 4A). To verify if the increase of ROS induced by
melatonin has an effect on cell viability, a known ROS scavenger, N-acetyl-L-cysteine (NAC, 10 mM),
was
 cellsused.  Figure
       did not         4Bbetween
                  differ  shows that
                                   the the  antioxidant
                                       vehicle          agent reverts
                                                groups (ethanol 0.3% the
                                                                      and10.9%);
                                                                           mM melatonin-induced
                                                                                 therefore, the meanviability
                                                                                                     of these
reduction
 rates was and     about 40%
             considered    as athe  effect
                                single     of melatonin 3 mM.
                                        group.

      Figure 4. Melatonin increases reactive oxygen species (ROS) production—(A) U87MG cells were
     Figure 4. Melatonin increases reactive oxygen species (ROS) production—(A) U87MG cells were
      incubated with melatonin (1 mM or 3 mM) for 72 h, and the medium was exchanged every 24 h.
     incubated with melatonin (1 mM or 3 mM) for 72 h, and the medium           was exchanged every 24 h. ROS
      ROS production was assessed by cytometry using the Muse® Cell Oxidative Stress kit. The results are
      presented as the percentage of cells positively labeled for superoxide radicals. ROS production and
      cell viability did not differ in cells treated with vehicle or 0.3% and 0.9% ethanol, and these groups are
      consequently represented as a single bar. (B) U87MG cell were incubated with vehicle melatonin (1 mM
      and 3 mM) and N-acetyl-L-cysteine (NAC, 10 mM) for 72 h and the medium was exchanged every 24 h.
      Proliferation was assessed based on the reaction with PrestoBlue (Thermo Fisher Scientific), and the
      fluorescence was read on a GloMax® 96 Microplate Luminometer (Promega Corporation). The results
      were presented as a percentage of the control group. As the proliferation did not differ between the
      vehicle-treated groups, they were represented as a single bar. * p < 0.05 compared to vehicle/control,
      # p < 0.05 compared to the group treated with 1 mM melatonin, and + p < 0.05 compared to the group
      treated with 3 mM melatonin. The statistical analysis consisted of an ANOVA followed by Bonferroni’s
Molecules 2018, 23, x FOR PEER REVIEW                                                                         6 of 16
      post-hoc test.

                                                   Figure 5. Cont.
Molecules 2018, 23, 1129                                                                                       6 of 16

Figure 5.Figure
          Melatonin      induces
                5. Melatonin         mitochondrial
                                induces   mitochondrial  membrane       depolarization
                                                           membrane depolarization      andand     apoptosis—U87MG
                                                                                             apoptosis—U87MG        cells cells
         were incubated    with  melatonin    (1 mM  or 3  mM)  for 72 h, and  the medium   was
were incubated with melatonin (1 mM or 3 mM) for 72 h, and the medium was exchanged every 24 h.   exchanged   every 24 h.
         (A) Mitochondrial polarization was evaluated by cytometry using a Muse® Mitopotential Assay Kit.
(A) Mitochondrial polarization was evaluated by cytometry using a Muse® Mitopotential Assay Kit.
         The depolarized group was represented by the sum of depolarized live and dead cells. Membrane
The depolarized    group
         polarization did notwas    represented
                                 differ              by with
                                        in cells treated  the sum    ofordepolarized
                                                               vehicle     0.3% and 0.9%  live  and and
                                                                                            ethanol,   dead  cells.
                                                                                                           these     Membrane
                                                                                                                 groups
polarization
         weredid   not differ
              represented         in cells
                            as a single  bar.treated   with was
                                               (B) Apoptosis  vehicle   or 0.3%
                                                                  evaluated         and 0.9%
                                                                              by cytometry       ethanol,
                                                                                             using  the Museand
                                                                                                             ®    these groups
                                                                                                                Annexin
         V & Dead   Cell  Assay  Kit. The  necrotic group   represents the sum   of dead cells and
were represented as a single bar. (B) Apoptosis was evaluated by cytometry using the Muse® Annexin  late apoptotic/dead
           cells presented in the representative plots. * p > 0.05 compared to vehicle. The statistical analysis
           consisted of an ANOVA followed by Bonferroni’s post-hoc test.
Molecules 2018, 23, x FOR PEER REVIEW                                                                                      7 of 16

     V & Dead Cell Assay Kit. The necrotic group represents the sum of dead cells and late apoptotic/dead
     cells presented in the representative plots. * p > 0.05 compared to vehicle. The statistical analysis
Molecules 2018, 23,
     consisted   of 1129
                    an ANOVA followed by Bonferroni’s post-hoc test.                                       7 of 16

2.6. Melatonin Arrested U87MG Cells at the G0/G1 Phase of the Cell Cycle
 2.6. Melatonin Arrested U87MG Cells at the G0/G1 Phase of the Cell Cycle
      In addition to the effect of melatonin on apoptosis, we investigated its ability to alter the cell
cycle.InAccordingly,
          addition to the effectcells
                      U87MG      of melatonin    on apoptosis,
                                      were incubated              we investigated
                                                         with either   1 mM or 3 its
                                                                                   mMability to alter
                                                                                        melatonin   forthe
                                                                                                         72cell
                                                                                                            h.
 cycle.  Accordingly,  U87MG     cells were   incubated   with   either 1 mM   or 3 mM   melatonin
Melatonin increased G0/G1 cell cycle arrest in U87MG cells at both 1 mM (71.92 ± 2.47%) and 3 mM     for  72 h.
 Melatonin
(77.77       increased
        ± 2.73%),      G0/G1
                  compared      cell vehicle
                             to the  cycle arrest
                                             groupin (64.85
                                                     U87MG    cells at(Figure
                                                            ± 1.20%)          6). (71.92 ± 2.47%) and 3 mM
                                                                       both 1 mM
 (77.77 ± 2.73%), compared to the vehicle group (64.85 ± 1.20%) (Figure 6).

                  Melatoninslows
      Figure6.6. Melatonin
     Figure                   slowsthethetransition
                                          transitionfrom
                                                       from thethe G0/G1
                                                                   G0/G1totoSSphase
                                                                                 phaseof ofthe
                                                                                            thecell
                                                                                                cell cycle—U87MG
                                                                                                      cycle—U87MGcells  cells
      were incubated
     were   incubated with
                         with melatonin
                              melatonin (1 (1 mM
                                              mM or or 33 mM)
                                                          mM) forfor 72
                                                                     72 h,
                                                                        h,and
                                                                           andthe
                                                                                themedium
                                                                                    mediumwas  wasexchanged
                                                                                                     exchangedeveryevery2424h.
      The  cell                                                                        ®
     h. The  cellcycle
                   cyclephases
                         phaseswere
                                 wereevaluated
                                        evaluatedby bycytometry
                                                         cytometryusing
                                                                      usingthe
                                                                            theMuse
                                                                                 Muse® Cell
                                                                                          Cell Cycle   Assay Kit.
                                                                                               Cycle Assay     Kit. The
                                                                                                                    Thecell
                                                                                                                         cell
      cycle distribution  did  not differ in cells treated   with  vehicle or 0.3%  and   0.9% ethanol,
     cycle distribution did not differ in cells treated with vehicle or 0.3% and 0.9% ethanol, and these  and  these groups
      were represented
     groups               as a single
              were represented      asbar.  * p > 0.05
                                        a single   bar.compared
                                                          * p > 0.05tocompared
                                                                        vehicle. The
                                                                                   to statistical analysis
                                                                                       vehicle. The         consisted
                                                                                                       statistical     of an
                                                                                                                   analysis
      ANOVA followed
     consisted             by Bonferroni’s
                  of an ANOVA     followed by post-hoc     test. post-hoc test.
                                                 Bonferroni’s

 2.7.Melatonin
2.7. MelatoninPotentiated
               Potentiatedthe
                           theEffect
                              EffectofofTemozolomide
                                        Temozolomide (TMZ)
                                                     (TMZ) to
                                                           to Reduce
                                                              Reduce Cell
                                                                     Cell Viability
                                                                          Viability
       To determine
       To determinethe  thepotential
                            potentialsynergistic
                                        synergisticeffect
                                                       effectof
                                                              ofmelatonin
                                                                 melatoninand  andthe
                                                                                    thechemotherapeutic
                                                                                        chemotherapeuticdrug  drugTMZ,
                                                                                                                    TMZ,
 we incubated
we    incubated U87MG
                  U87MG cellscellsfor
                                    for 72
                                         72 hh with
                                               with 11 mMmM oror 33 mM
                                                                     mM of of melatonin
                                                                              melatonin in in combination
                                                                                               combination withwith TMZ
                                                                                                                    TMZ
 (0.8 mM).
(0.8  mM). Tumor
             Tumor cell viability
                          viability was
                                      was measured
                                            measuredby    bydetecting
                                                             detectingthethefluorescence
                                                                             fluorescenceemitted
                                                                                             emittedbybythe
                                                                                                          thereduction
                                                                                                               reductionof
 the  PrestoBlue  ® reagent   by  living   cells.  The  results obtained    showed    that 1 mM   and  3 mM    melatonin
of the PrestoBlue reagent by living cells. The results obtained showed that 1 mM and 3 mM
                      ®

 reduced cell
melatonin       viability/proliferation
              reduced                        by 10% and by
                         cell viability/proliferation       34%,10%respectively.
                                                                       and 34%,TMZ       reduced cell
                                                                                    respectively.  TMZ viability
                                                                                                           reducedby 45%.
                                                                                                                     cell
 Therefore,
viability  bythe addition
               45%.          of 1 mM
                     Therefore,    the and   3 mMofmelatonin
                                        addition        1 mM and  increased   the effect by
                                                                     3 mM melatonin          49% andthe
                                                                                          increased   87%,   respectively
                                                                                                          effect by 49%
 (Figure
and   87%,7).  Cells treated
           respectively        with7).
                           (Figure   vehicles     (ethanol
                                         Cells treated      0.3%
                                                          with     or 0.9%,
                                                                vehicles      dimethylsulfoxide
                                                                          (ethanol                  (DMSO) 0.1%, or a
                                                                                     0.3% or 0.9%, dimethylsulfoxide
 combination
(DMSO)     0.1%,thereof)   are represented
                  or a combination              as aare
                                          thereof)    single bar in Figure
                                                          represented    as a 6single
                                                                                becausebarthese  vehicles
                                                                                            in Figure      did not these
                                                                                                       6 because    affect
 cell viability and  the  viability  did   not significantly    differ between    these  groups.
vehicles did not affect cell viability and the viability did not significantly differ between these groups.
Molecules
 Molecules2018,
           2018,23,
                 23,x1129
                      FOR PEER REVIEW                                                                                8 8ofof16
                                                                                                                             16

                  Melatoninpotentiates
      Figure7.7. Melatonin
     Figure                   potentiatesthe
                                           theeffect
                                               effectof
                                                      oftemozolomide
                                                         temozolomide(TMZ) (TMZ)on on cell
                                                                                       cell proliferation/survival—
                                                                                            proliferation/survival—
     U87MG cells were incubated with melatonin (1 mM or 3 mM) in combination not
      U87MG     cells were incubated with  melatonin  (1 mM    or 3 mM)   in combination   or     withwith
                                                                                               or not   TMZ  (0.8 mM)
                                                                                                            TMZ   (0.8
      for 72 h, and   the medium  was  exchanged    every  24 h.  Proliferation was  assessed   based
     mM) for 72 h, and the medium was exchanged every 24 h. Proliferation was assessed based on the    on the reaction
      with PrestoBlue    (Thermo(Thermo
                                 Fisher Scientific), and the fluorescence     was read on  a GloMax   ®
     reaction with PrestoBlue               Fisher Scientific), and the fluorescence    was   read on a 96  Microplate
                                                                                                         GloMax  ® 96

     Microplate Luminometer (Promega Corporation). The results are presented as a percentage of theof
      Luminometer       (Promega  Corporation).    The  results  are presented   as  a percentage   of  the vehicle
      each group. Proliferation did not differ between the vehicle-treated groups, and these groups were
     vehicle of each group. Proliferation did not differ between the vehicle-treated groups, and these
      represented as a single bar. * p < 0.05 compared to vehicle, # p < 0.05 compared to the group treated with
     groups were represented as a single bar. * p < 0.05 compared to vehicle, # p < 0.05 compared to the
      1 mM melatonin, and + p < 0.05 compared to the group treated with 3 mM melatonin. The statistical
     group treated with 1 mM melatonin, and + p < 0.05 compared to the group treated with 3 mM
      analysis consisted of an ANOVA followed by Bonferroni’s post-hoc test.
     melatonin. The statistical analysis consisted of an ANOVA followed by Bonferroni’s post-hoc test.

3.3.Discussion
     Discussion
       Mitochondriaare
      Mitochondria      arecentral
                             centralorganelles
                                       organellesininthe
                                                       thedevelopment
                                                             developmentofofcancer
                                                                               cancerbecause
                                                                                         becausethey
                                                                                                   theyare
                                                                                                         areresponsible
                                                                                                               responsible
 forthe
for  thebalance
          balanceof ofbioenergetic
                       bioenergeticand  andbiosynthetic
                                              biosyntheticprocesses,
                                                              processes,and
                                                                         andbecause
                                                                               becausethey theyare
                                                                                                 arethe
                                                                                                     themain
                                                                                                          mainsource
                                                                                                                  sourceof of
 superoxides    and   are  implicated     in  the intrinsic   apoptosis  pathway.     Besides    that,
superoxides and are implicated in the intrinsic apoptosis pathway. Besides that, the transcription     the   transcription
 factorTFAM
factor  TFAMisisessential
                   essentialforforthe
                                    thereplication,
                                        replication,transcription,
                                                      transcription,andandmaintenance
                                                                             maintenanceofofmtDNA
                                                                                                mtDNAand,  and,therefore,
                                                                                                                  therefore,
 formitochondrial
for  mitochondrial homeostasis
                      homeostasis [8–10].
                                       [8–10]. Recent
                                                Recent discoveries
                                                         discoverieshave
                                                                      haveshown
                                                                             shownthatthatmitochondria
                                                                                            mitochondria   arearea target for
                                                                                                                    a target
 melatonin   [28], and  melatonin     has   been shown    to accumulate   in mitochondria
for melatonin [28], and melatonin has been shown to accumulate in mitochondria against the     against the  concentration
 gradient via active
concentration    gradienttransport    [29].transport
                             via active       This phenomenon       remains poorly
                                                      [29]. This phenomenon            understood,
                                                                                   remains              but melatonin
                                                                                               poorly understood,        butis
 known    to prevent   the  inhibition   of  complexes    I and  IV induced   by  red  ruthenium
melatonin is known to prevent the inhibition of complexes I and IV induced by red ruthenium [30].    [30].  In  the  present
 study,
In       we showed
   the present   study,that
                          weTFAM
                               showed andthat
                                            other mitochondrial
                                                TFAM     and othertranscription
                                                                     mitochondrial factors   (TFB1M and
                                                                                       transcription          TFB2M)
                                                                                                        factors    (TFB1Mmay
 be targets
and  TFB2M)ofmay melatonin     in glioblastoma
                      be targets   of melatonin in  cells.  The melatonin-induced
                                                       glioblastoma                       reduction in the
                                                                       cells. The melatonin-induced             expression
                                                                                                             reduction    in
the expression of these transcription factors was associated with reduced mitochondrial NADH1
 of these  transcription    factors   was    associated   with  reduced   mitochondrial      NADH      dehydrogenase
 (MT-ND1) gene1 expression.
dehydrogenase        (MT-ND1) gene   In accordance
                                           expression.with     our results,with
                                                         In accordance       Prunet-Marcassus       and colleaguesand
                                                                                our results, Prunet-Marcassus            [31]
 also  showed   that   melatonin     reduces    the transcriptional    content  of  mitochondria
colleagues [31] also showed that melatonin reduces the transcriptional content of mitochondria by      by  44%    in  brown
 adipocytes
44%  in brown in adipocytes
                 the Siberianinhamster.
                                   the Siberian hamster.
       Evidenceindicates
      Evidence    indicatesthat
                              thatTFAM
                                     TFAMhas  hasbeen
                                                  beenshown
                                                        shownto  toregulate
                                                                    regulatemtDNA
                                                                              mtDNAcopy  copynumber
                                                                                                number[10,11],
                                                                                                          [10,11],and andanan
 unbalance   in the  number     of mtDNA       copies is associated   with  several  neurodegenerative
unbalance in the number of mtDNA copies is associated with several neurodegenerative diseases and             diseases   and
 cancer,including
cancer,   includingglioblastoma
                      glioblastoma[13,32–34].
                                        [13,32–34].Melatonin
                                                      Melatonindid didnot
                                                                       notaffect
                                                                            affectthe
                                                                                   themtDNA
                                                                                        mtDNAcopy  copynumber
                                                                                                          numberininthis this
 study, despite
study,   despite reducing
                  reducing mitochondrial
                              mitochondrialgene  geneexpression,
                                                        expression,which
                                                                      whichindicates
                                                                              indicates that  different
                                                                                           that differentmechanisms
                                                                                                            mechanisms    are
 responsible   for  the control    of mitochondrial      gene   expression   and mtDNA        replication
are responsible for the control of mitochondrial gene expression and mtDNA replication by melatonin.        by   melatonin.
In cultures of mouse C6 gliomas and Neuro2a mice, melatonin reverses the morphine- and nickel
Molecules 2018, 23, 1129                                                                              9 of 16

In cultures of mouse C6 gliomas and Neuro2a mice, melatonin reverses the morphine- and nickel
chloride-induced reductions in mtDNA copy number, respectively. But, in accordance with our results,
melatonin alone does not interfere with mtDNA content [35,36].
      Changes in mitochondrial gene expression or nuclear genes may result in the collapse of the
mitochondrial respiratory chain to increase ROS production, which, consequently, may trigger the
activation of apoptosis [3–5,37–40]. Our results showed that melatonin increased ROS production,
which may be a consequence of alterations in the expression of respiratory chain genes that depend
on the activity of the transcription factors TFAM, TFB1M, and TFB2M. This hypothesis might be
supported by the fact that the downregulation of TFAM expression has promoted ROS-dependent
activation of JNK/p38 MAPK and apoptosis, as reported in non-small cell lung cancer [41]. In addition,
in the cardiac muscle cell line, the downregulation of TFAM caused mitochondrial oxidative
phosphorylation dysfunction, resulting in increased ROS production [42]. Still, the overexpression
of TFAM inhibited mitochondrial ROS generation in HeLa cells [43] and prevented oxidative stress,
facilitating cardioprotection [44].
      High concentrations of ROS can damage mtDNA and nuclear DNA and alter the expression of
oncogenes and tumor suppressor genes, which modifies the onset and progression of tumors [37].
Several studies demonstrated that melatonin influences the intracellular content of ROS by different
mechanisms. The protective role of melatonin by reducing oxidative stress in experimental models
of tissue damage is well known [24]. For example, in contrast to our results, in a non-cancerous
model of myocardial ischemia/reperfusion injury in type 1 diabetic rats, melatonin has increased
TFAM expression, reducing mitochondrial oxidative stress and enhancing its biogenesis [45]. However,
in cancer cells, two scenarios have been shown: A pro-oxidant activity in which melatonin induces the
increase of intracellular levels of ROS, leading to cell death, as was shown in the present study and
by others [26,46–54]; and melatonin reducing intracellular levels of ROS and inducing cell death by
different mechanisms, such as, for example, inhibiting the nuclear transcription factor kappa B (NF-κB)
nuclear activity, as reported in human glioma cells (T98 and U251) by Wang and colleagues [55],
and in rat glioma cells (C6) by Martín and colleagues [56]. Otherwise, the melatonin activation of
NF-κB has also been associated with an increase in intracellular oxidative stress in a model of human
monocyte (U937) culture [47,48], and in a primary cerebellar granule cell culture [57]. In the present
study, a known antioxidant, NAC, completely inhibited the decreased viability induced by 1 mM
melatonin in a U87MG cell culture. At a melatonin concentration of 3 mM, NAC inhibited this effect
by about 40%, indicating that melatonin-induced ROS increase is at least in part responsible for the
observed cell death. It is important to consider the concentration and the time of exposure to melatonin.
The pharmacological concentrations of melatonin used (1 mM and 3 mM) indicate that the observed
effects are independent of melatonin receptors (MT1 and MT2). Minor concentrations of melatonin
(1 µM and 100 µM) had no effect on cell proliferation or on the expression of TFAM (data not shown).
The concentration of 3 mM of melatonin presented a more consistent result regarding the reduction of
TFAM at the protein level. Although not statistically significant, a trend of TFAM protein decrease
was also observed at the concentration of 1 mM (supplemental material). Interestingly, 1 mM of
melatonin was enough to alter the mitochondrial gene (MT-ND1) expression. The exposition time to
melatonin also proved to be crucial for the observed results. Incubation times with melatonin of less
than 72 h were not sufficient to alter cell viability, as well as intracellular ROS content (data not shown).
Wang and colleagues [55] have also shown no change in cell proliferation after 24 h of incubation with
melatonin, although they have detected a decrease in intracellular ROS levels.
      Our results showed that mitochondrial membrane depolarization was increased in cells incubated
with melatonin, which indicates a collapse of inner membrane polarization that triggers the opening
of mitochondrial transition pores (MTPs) and the release of cytochrome C and other pro-apoptotic
factors [28,38,58,59]. The increase in ROS production induced by melatonin might lead to mitochondrial
membrane depolarization and the activation of cell death, although confirmation of the activation of
intrinsic apoptosis needs to be proven. Melatonin is known to affect MTP in non-cancer cells, such as
Molecules 2018, 23, 1129                                                                            10 of 16

in striatal neurons. In this model, melatonin prevents loss of mitochondrial membrane potential and
reduces the probability of MTP opening, which prevents cell death by apoptosis [60]. Whereas in
human promyelocytic leukaemia HL-60 cells, melatonin increases H2 O2 -induced ROS generation,
causing a decrease in mitochondrial membrane potential and cell death [51]. The opposing effects of
melatonin on cancer and non-cancer cells has been widely discussed and can be reviewed in [23,26].
      In addition to the effects on oxidative stress and cell death, we showed that melatonin inhibits
the progression of the cell cycle in U87MG cells. A possible pathway to explain this cell cycle arrest
is through the physical interaction between the tumor suppressor protein p53 and TFAM [61–64].
Furthermore, p53 is a target of melatonin, which activates p53, and in its turn induces apoptosis and
arrests tumor cells in the G1/G0 to S transition of the cell cycle [26,65].
      Glioblastoma is a very aggressive type of cancer with a very low survival rate. Therefore, new
therapeutic targets have been investigated, and TFAM is a strong candidate target since its expression
is altered in several types of cancer, including glioma [13,15], colorectal cancer [63], epithelial ovarian
carcinoma [66], bladder cancer [67], breast cancer [68], lung cancer [41,69], and colon cancer [70].
Moreover, the use of melatonin in the treatment of cancer has shown promising results. Specifically,
previous studies have demonstrated a possible antitumor role for melatonin in glioma models [47,48].
In addition, Kinker and colleagues [71] have recently demonstrated that human glioma cell lines (HOG,
T98G, and U87MG) produce melatonin, and the ability of cells to produce this hormone negatively
correlated with tumor malignancy.
      Finally, our results showed that the combination of melatonin with temozolamide, TMZ,
potentiated its effects on cell survival, pointing at a promising combinatorial treatment for glioblastoma
patients. In summary, our results suggest that increased generation of melatonin-induced intracellular
ROS in U87MG glioblastoma cells may be an effect of melatonin on the expression of TFAM and
other mitochondrial transcription factors (TFB1M and TFB2M), leading to mitochondrial disruption.
Our study opens a new perspective to understand the mechanism of action of melatonin in tumor cells.

4. Materials and Methods

4.1. Cell Culture Conditions
     The human malignant glioma cell line U87MG (American Type Culture Collection, ATCC) was
routinely cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Life Technologies, Carlsbad, CA,
USA), supplemented with 10% fetal bovine serum (FBS) (Life Technologies), 100 IU/mL penicillin, and
100 µg/mL streptomycin (Life Technologies), in a humidified atmosphere consisting of 5% CO2 in air
at 37 ◦ C. The cell line was authenticated by short tandem repeat DNA profiling using the GenePrint 10
System (Promega, Madison, WI, USA).

4.2. Extraction of RNA and DNA
     Cells were plated 2 × 105 cells/mL in a 24-well plate and treated with 1 mM or 3 mM of melatonin
for 72 h, and the control groups were treated with 0.3% or 0.9% of ethanol vehicle, respectively.
The medium containing melatonin or vehicle was changed every 24 h. The cells were then digested with
RLT Plus buffer (QIAGEN, Hilden, Germany), syringe homogenized 10 times, and frozen at −80 ◦ C
before extracting the genetic material. Both RNA and DNA were extracted from the cell homogenate
using the AllPrep DNA/RNA Micro Kit (QIAGEN) following the protocol provided by the manufacturer.
The concentration (ng/µL) and purity of DNA and total RNA were determined by quantification on the
NanoDrop ND-1000 spectrophotometer (Thermo Scientific, Wilmington, DE, USA).

4.3. Expression of TFAM, TFB1M, TFB2M, and NADH Dehydrogenase 1 (MT-ND1) by qRT-PCR
    RNA was reverse transcribed using the Maxima First Strand cDNA Synthesis kit for qRT-PCR
(Thermo Scientific) according to the manufacturer’s specifications. Quantitative data were obtained
using SYBR green (Thermo Scientific) qRT-PCR on the ABI Prism 7500 sequence detector (Applied
Molecules 2018, 23, 1129                                                                              11 of 16

Biosystems, Foster City, CA, USA), and normalized in relation to the geometric mean of three
housekeeping genes: Hypoxanthime phosphoribosyltransferase (HPRT), glucuronidase beta (GUSB),
and TATA binding protein (TBP). The equation 2−∆∆Ct was applied to calculate the relative gene
expression levels [27]. The primers were designed to amplify 80–150 bp length amplicons, had a melting
temperature of 60 ◦ C, and were synthesized by Integrate DNA Technology (IDT, Coralville, IA, USA)
as follows (50 to 30 ); TFAM F: CTCCCCCTTCAGTTTTGTGT, TFAM R: GCATCGGG-TTCTGAGCTTT;
TFB1M F: ATGGCTCAGTACCTCTGCAATG, TFB1M R: TGGGCTGTATCAAGGGAGTGA; TFB2M F:
ATCCCGGAAATCCAGACTTGT, TFB2M R: GACCAAGGCTCCATGTGCA; NADH dehydrogenase
1 (MT-ND1) F: TGATGGCTAGGGTGACTTCAT, MT-ND1 R: CCTAGCCGTTTACTCAATCCT;
HPRT F: TGAGGATTTGGAAAGGGTGT, HPRT R: GAGCACACAGAGGGCTACAA; GUSB F:
GAAAATACGTGGTTGGAGAGCTCATT, GUSB R: CCGAGTGAAGATCCCCTTTTTA; TBP F:
AGGATAAGAGAGCCACGAACCA, TBP R: CTTGCTGCCAGTCTGGACTGT. PCR was carried out
as follows: 5 min at 50 ◦ C, 10 min at 95 ◦ C, 40 cycles at 95 ◦ C for 15 s, and 60 ◦ C for 1 min. The primer
concentrations used were 200–400 nM. All assays were carried out in duplicate and eventually repeated
when the standard deviation exceeded 0.4.

4.4. Mitochondrial DNA Copy Number Quantification
      A single copy gene—hemoglobin beta (HBB)—was used as a reference to determine the number
of copies of mtDNA by SYBR Green qRT-PCR on an ABI Prism 7500 sequence detector (Applied
Biosystems). The primer sequence used to quantify the mtDNA copy number was the same as
that of NADH dehydrogenase 1, and the primer was used at a final concentration of 200 nM.
The sequences of HBB were as follows (50 –30 ): HBB F: GTGAAGGCTCATGGCAAGA and HBB
R: AGCTCACTCAGGTGTGGCAAAG (IDT). The cycle conditions were 10 min at 95 ◦ C, 40 cycles at
95 ◦ C for 15 s, and 60 ◦ C for 1 min. All assays were carried out in duplicate and eventually repeated
when the standard deviation exceeded 0.4. The relative mtDNA copy number was determined with
the equation 2−∆∆Ct [27].

4.5. Western Blot Analysis
     Total protein lysates were prepared from U98MG cell cultures with RIPA lysis buffer and protease
inhibitor cocktail (Sigma-Aldrich) on ice. The protein concentration was determined using a NanoDrop
Microvolume Spectrophotometers (Thermo Scientific™). Total protein lysates (30 mg) were separated
by 4% to 12% polyacrylamide gel electrophoresis (Invitrogen, Carsbald, CA, USA) with 1× NuPAGE
MOPS SDS 20× (Invitrogen, Carsbald, CA, USA) running buffer. The proteins were electrophoretically
transferred to a Polyvinylidene Fluoride membrane (PVDF) through the semi-dry Trans-Blot® SD
system (Trans-Blot® Transfer Cell, Biorad, Hercules, CA, USA). The membrane was blocked with
5% skim milk and incubated with rabbit monoclonal primary anti-TFAM diluted 1:500, and with
mouse monoclonal primary anti-β-actina (clone AC-74, Sigma-Aldrich) diluted 1:5000, as a protein
loading control. The secondary antibodies used were anti-rabbit (1:1000) and anti-mouse IgG (1:5000)
conjugated to peroxidase (Sigma-Aldrich). The immune complexes were visualized using enhanced
chemiluminescence reagent (Western Lightning Chemiluminescence Reagent Plus, Perkin Elmer,
Waltham, MA, USA) and detected with UVITEC (Alliance 4.7) Cambridge, UK.

4.6. Evaluation of Oxidative Stress, Cell Cycle, Apoptosis, and Mitochondria Polarization
     U87MG cells were treated with 1 mM or 3 mM melatonin for 72 h. Control groups were treated
with ethanol vehicle (0.3% or 0.9%, relative to the melatonin concentration). The medium was changed
every 24 h, and the melatonin or vehicle was replaced. The cells were then prepared for cytometric
assays to evaluate oxidative stress, the cell cycle, apoptosis, and mitochondrial membrane polarization
using the Muse® Cell Analyzer (Merck Millipore, Billerica, MA, USA) and appropriate reagent kits:
Muse® Cell Oxidative Stress Kit, Muse® Cell Cycle Assay Kit, Muse® Annexin V & Dead Cell Assay Kit
and Muse® Mitopotential Assay Kit, respectively, according to the manufacturer’s instructions.
Molecules 2018, 23, 1129                                                                                       12 of 16

4.7. Cell Viability/Proliferation
     U87MG cells were treated with melatonin (1 mM or 3 mM) in the presence or absence of
temozolomide (TMZ 0.8 mM—Sigma-Aldrich, St. Louis, MO, USA) or N-Acetyl-L-cysteine (NAC 10 mM—
Sigma-Aldrich, St. Louis, MO, USA) for 72 h, and the medium and drug were replaced every
24 h. The control groups were treated with melatonin vehicle (0.3% or 0.9% ethanol), TMZ vehicle
(0.1% dimethylsulfoxide—DMSO), or a combination thereof (0.3% ethanol + DMSO 0.1% or 0.9% ethanol
+ 0.1% DMSO). NAC was diluted in DMEM. After the treatments, the cells were incubated with PrestoBlue
reagent (Invitrogen, Carlsbad, CA, USA) for 2 h, and the fluorescence was measured on a GloMax® 96
Microplate Luminometer (Promega Corporation, Madison, WI, USA).

4.8. Statistical Analysis
     The results are reported as the mean ± s.e.m. of at least three independent experiments, and were
normalized to the groups treated with specific vehicles for each experiment. The differences between
experimental groups were tested with an analysis of variance followed by the Bonferroni post-hoc
correction using GraphPad Prism® version 5.

Supplementary Materials: The following are available online.
Author Contributions: D.G.F. and S.K.N.M. conceived, designed the experiments and contributed with the
analysis of the data; D.G.F. and I.F.M. performed the experiments; All authors critically revised the manuscript.
Funding: This research was funded by Fundação de Amparo a Pesquisa de São Paulo (FAPESP) grant
numbers [#2004/12133-6, #2013/02162-8, #2014/17220-6] and Conselho Nacional de Desenvolvimento Científico
e Tecnológico (CNPq) grant number [#305730/2015-0].
Conflicts of Interest: The authors declare no conflict of interest.

References
1.    Nass, N.M. The circularity of Mitochondrial DNA. Proc. Natl. Acad. Sci. USA 1966, 56, 1215–1222. [CrossRef]
      [PubMed]
2.    Van Bruggen, E.F.; Borst, P.; Ruttenberg, G.J.; Gruber, M.; Kroon, A.M. Circular mitochondrial DNA.
      Biochim. Biophys. Acta 1966, 119, 437–439. [CrossRef]
3.    Kauppila, T.E.S.; Kauppila, J.H.K.; Larsson, N.G. Mammalian Mitochondria and Aging: An Update.
      Cell Metab. 2016, 25, 57–71. [CrossRef] [PubMed]
4.    Baffy, G. Mitochondrial uncoupling in cancer cells: Liabilities andopportunities. Biochim. Biophys. Acta 2017,
      185, 655–664. [CrossRef] [PubMed]
5.    Trotta, A.P.; Chipuk, J.E. Mitochondrial dynamics as regulators of cancer biology. Cell. Mol. Life Sci. 2017, 74,
      1999–2017. [CrossRef] [PubMed]
6.    Falkenberg, M.; Larsson, N.G.; Gustafsson, C.M. DNA replication and transcription in mammalian
      mitochondria. Annu. Rev. Biochem. 2007, 76, 679–699. [CrossRef] [PubMed]
7.    Bogenhagen, D.F. Mitochondrial DNA nucleoid structure. Biochim. Biophys. Acta 2012, 1819, 914–920.
      [CrossRef] [PubMed]
8.    Alam, T.I.; Kanki, T.; Muta, T.; Ukaji, K.; Abe, Y.; Nakayama, H.; Takio, K.; Hamasaki, N.; Kang, D. Human
      mitochondrial DNA is packaged with TFAM. Nucleic Acids Res. 2003, 31, 1640–1645. [CrossRef] [PubMed]
9.    Kanki, T.; Nakayama, H.; Sasaki, N.; Takio, K.; Alam, T.I.; Hamasaki, N.; Kang, D. Mitochondrial nucleoid
      and transcription factor A. Ann. N. Y. Acad. Sci. 2004, 1011, 61–68. [CrossRef] [PubMed]
10.   Campbell, C.T.; Kolesar, J.E.; Kaufman, B.A. Mitochondrial transcription factor A regulates mitochondrial
      transcription initiation, DNA packaging, and genome copy number. Biochim. Biophys. Acta 2012, 1819,
      921–929. [CrossRef] [PubMed]
11.   Ekstrand, M.I.; Falkenberg, M.; Rantanen, A.; Park, C.B.; Gaspari, M.; Hultenby, K.; Rustin, P.;
      Gustafsson, C.M.; Larsson, N.G. Mitochondrial transcription factor A regulates mtDNA copy number
      in mammals. Hum. Mol. Genet. 2004, 13, 935–944. [CrossRef] [PubMed]
Molecules 2018, 23, 1129                                                                                        13 of 16

12.   Lin, P.C.; Lin, J.K.; Yang, S.H.; Wang, H.S.; Li, A.F.; Chang, S.C. Expression of beta-F1-ATPase and
      mitochondrial transcription factor A and the change in mitochondrial DNA content in colorectal cancer:
      Clinical data analysis and evidence from an in vitro study. Int. J. Colorectal. Dis. 2008, 23, 1223–1232.
      [CrossRef] [PubMed]
13.   Correia, R.L.; Oba-Shinjo, S.M.; Uno, M.; Huang, N.; Marie, S.K. Mitochondrial DNA depletion and
      its correlation with TFAM, TFB1M, TFB2M and POLG in human diffusely infiltrating astrocytomas.
      Mitochondrion 2011, 11, 48–53. [CrossRef] [PubMed]
14.   Yoshida, Y.; Hasegawa, J.; Nezu, R.; Kim, Y.K.; Hirota, M.; Kawano, K.; Izumi, H.; Kohno, K. Clinical
      usefulness of mitochondrial transcription factor A expression as a predictive marker in colorectal cancer
      patients treated with FOLFOX. Cancer Sci. 2011, 102, 578–582. [CrossRef] [PubMed]
15.   Jiang, J.; Yang, J.; Wang, Z.; Wu, G.; Liu, F. TFAM is directly regulated by miR-23b in glioma. Oncol. Rep.
      2013, 30, 2105–2110. [CrossRef] [PubMed]
16.   Omuro, A.; DeAngelis, L.M. Glioblastoma and other malignant gliomas: A clinical review. JAMA 2013, 310,
      1842–1850. [CrossRef] [PubMed]
17.   Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.;
      Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of
      the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820. [CrossRef] [PubMed]
18.   Mineo, J.F.; Bordron, A.; Baroncini, M.; Ramirez, C.; Maurage, C.A.; Blond, S.; Dam-Hieu, P. Prognosis
      factors of survival time in patients with glioblastoma multiforme: A multivariate analysis of 340 patients.
      Acta Neurochir. 2007, 149, 245–252. [CrossRef] [PubMed]
19.   Furnari, F.B.; Fenton, T.; Bachoo, R.M.; Mukasa, A.; Stommel, J.M.; Stegh, A.; Hahn, W.C.; Ligon, K.L.;
      Louis, D.N.; Brennan, C.; et al. Malignant astrocytic glioma: Genetics, biology, and paths to treatment.
      Genes Dev. 2007, 21, 2683–2710. [CrossRef] [PubMed]
20.   Cloughesy, T.F.; Cavenee, W.K.; Mischel, P.S. Glioblastoma: From molecular pathology to targeted treatment.
      Annu. Rev. Pathol. 2014, 9, 1–25. [CrossRef] [PubMed]
21.   Parker, N.R.; Correia, N.; Crossley, B.; Buckland, M.E.; Howell, V.M.; Wheeler, H.R. Correlation of MicroRNA
      132 Up-regulation with an Unfavorable Clinical Outcome in Patients with Primary Glioblastoma Multiforme
      Treated with Radiotherapy Plus Concomitant and Adjuvant Temozolomide Chemotherapy. Transl. Oncol.
      2013, 6, 742–748. [CrossRef] [PubMed]
22.   Ichimura, K.; Narita, Y.; Hawkins, C.E. Diffusely infiltrating astrocytomas: Pathology, molecular mechanisms
      and markers. Acta Neuropathol. 2015, 129, 789–808. [CrossRef] [PubMed]
23.   Sainz, R.M.; Mayo, J.C.; Rodriguez, C.; Tan, D.X.; Lopez-Burillo, S.; Reiter, R.J. Melatonin and cell death:
      Differential actions on apoptosis in normal and cancer cells. Cell. Mol. Life Sci. 2003, 60, 1407–1426. [CrossRef]
      [PubMed]
24.   Luchetti, F.; Canonico, B.; Betti, M.; Arcangeletti, M.; Pilolli, F.; Piroddi, M.; Canesi, L.; Papa, S.; Galli, F.
      Melatonin signaling and cell protection function. FASEB J. 2010, 24, 3603–3624. [CrossRef] [PubMed]
25.   Radogna, F.; Nuccitelli, S.; Mengoni, F.; Ghibelli, L. Neuroprotection by melatonin on astrocytoma cell death.
      Ann. N. Y. Acad. Sci. 2009, 1171, 509–513. [CrossRef] [PubMed]
26.   Lanoix, D.; Lacasse, A.A.; Reiter, R.J.; Vaillancourt, C. Melatonin: The smart killer: The human trophoblast as
      a model. Mol. Cell. Endocrinol. 2012, 348, 1–11. [CrossRef] [PubMed]
27.   Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and
      the 2(-Delta Delta C(T)). Method Methods 2001, 25, 402–408. [CrossRef] [PubMed]
28.   Acuña Castroviejo, D.; Escames, G.; Carazo, A.; León, J.; Khaldy, H.; Reiter, R.J. Melatonin, mitochondrial
      homeostasis and mitochondrial-related diseases. Curr. Top Med. Chem. 2002, 2, 133–151. [CrossRef] [PubMed]
29.   Tan, D.X.; Manchester, L.C.; Qin, L.; Reiter, R.J. Melatonin: A Mitochondrial Targeting Molecule Involving
      Mitochondrial Protection and Dynamics. Int. J. Mol. Sci. 2016, 17, 2124. [CrossRef] [PubMed]
30.   Martín, M.; Macías, M.; Escames, G.; Reiter, R.J.; Agapito, M.T.; Ortiz, G.G.; Acuña-Castroviejo, D.
      Melatonin-induced increased activity of the respiratory chain complexes I and IV can prevent mitochondrial
      damage induced by ruthenium red in vivo. J. Pineal Res. 2000, 28, 242–248. [CrossRef] [PubMed]
31.   Prunet-Marcassus, B.; Ambid, L.; Viguerie-Bascands, N.; Pénicaud, L.; Casteilla, L. Evidence for a direct
      effect of melatonin on mitochondrial genome expression of Siberian hamster brown adipocytes. J. Pineal Res.
      2001, 30, 108–115. [CrossRef] [PubMed]
Molecules 2018, 23, 1129                                                                                             14 of 16

32.   Guo, J.; Zheng, L.; Liu, W.; Wang, X.; Wang, Z.; Wang, Z.; French, A.J.; Kang, D.; Chen, L.; Thibodeau, S.N.;
      et al. Frequent truncating mutation of TFAM induces mitochondrial DNA depletion and apoptotic resistance
      in microsatellite-unstable colorectal cancer. Cancer Res. 2011, 71, 2978–2987. [CrossRef] [PubMed]
33.   Dickinson, A.; Yeung, K.Y.; Donoghue, J.; Baker, M.J.; Kelly, R.D.; McKenzie, M.; Johns, T.G.; St. John, J.C.
      The regulation of mitochondrial DNA copy number in glioblastoma cells. Cell Death Differ. 2013, 20,
      1644–1653. [CrossRef] [PubMed]
34.   Meng, S.; Han, J. Mitochondrial DNA copy number alteration in human cancers. N. Am. J. Med. Sci. 2013, 6,
      22–25. [CrossRef]
35.   Feng, Y.M.; Jia, Y.F.; Su, L.Y.; Wang, D.; Lv, L.; Xu, L.; Yao, Y.G. Decreased mitochondrial DNA copy number
      in the hippocampus and peripheral blood during opiate addiction is mediated by autophagy and can be
      salvaged by melatonin. Autophagy 2013, 9, 1395–1406. [CrossRef] [PubMed]
36.   Xu, S.C.; He, M.D.; Lu, Y.H.; Li, L.; Zhong, M.; Zhang, Y.W.; Wang, Y.; Yu, Z.P.; Zhou, Z. Nickel exposure
      induces oxidative damage to mitochondrial DNA in Neuro2a cells: The neuroprotective roles of melatonin.
      J. Pineal Res. 2011, 51, 426–433. [CrossRef] [PubMed]
37.   Thannickal, V.J.; Fanburg, B.L. Reactive oxygen species in cell signaling. Am. J. Physiol. Lung Cell. Mol. Physiol.
      2000, 279, L1005–L1028. [CrossRef] [PubMed]
38.   De Miguel, M.; Cordero, M.D. Oxidative Therapy against Cancer. In Oxidative Stress and Diseases;
      Lushchak, V.I., Gospodaryov, D.V., Eds.; InTech: Rijeka, Croatia, 2012; Available online: http://
      www.intechopen.com/books/oxidative-stress-and-diseases/oxidative-therapy-against-cancer (accessed
      on 4 April 2018).
39.   Wu, C.C.; Bratton, S.B. Regulation of the intrinsic apoptosis pathway by reactive oxygen species.
      Antioxid. Redox Signal. 2013, 19, 546–558. [CrossRef] [PubMed]
40.   Rinaldi, M.; Caffo, M.; Minutoli, L.; Marini, H.; Abbritti, R.V.; Squadrito, F.; Trichilo, V.; Valenti, A.; Barresi, V.;
      Altavilla, D.; et al. ROS and Brain Gliomas: An Overview of Potential and Innovative Therapeutic Strategies.
      Int. J. Mol. Sci. 2016, 17, 984. [CrossRef] [PubMed]
41.   Xie, D.; Wu, X.; Lan, L.; Shangguan, F.; Lin, X.; Chen, F.; Xu, S.; Zhang, Y.; Chen, Z.; Huang, K.; et al.
      Downregulation of TFAM inhibits the tumorigenesis of non-small cell lung cancer by activating ROS-
      mediated JNK/p38MAPK signaling and reducing cellular bioenergetics. Oncotarget 2016, 7, 11609–11624.
      [CrossRef] [PubMed]
42.   Kunkel, G.H.; Chaturvedi, P.; Tyagi, S.C. Mitochondrial pathways to cardiac recovery: TFAM. Heart Fail. Rev.
      2016, 21, 499–517. [CrossRef] [PubMed]
43.   Hayashi, Y.; Yoshida, M.; Yamato, M.; Ide, T.; Wu, Z.; Ochi-Shindou, M.; Kanki, T.; Kang, D.; Sunagawa, K.;
      Tsutsui, H.; et al. Reverse of age-dependent memory impairment and mitochondrial DNA damage in
      microglia by an overexpression of human mitochondrial transcription factor a in mice. J. Neurosci. 2008, 28,
      8624–8634. [CrossRef] [PubMed]
44.   Ikeda, M.; Ide, T.; Fujino, T.; Arai, S.; Saku, K.; Kakino, T.; Tyynismaa, H.; Yamasaki, T.; Yamada, K.; Kang, D.;
      et al. Overexpression of TFAM or twinkle increases mtDNA copy number and facilitates cardioprotection
      associated with limited mitochondrial oxidative stress. PLoS ONE 2015, 10, e0119687. [CrossRef] [PubMed]
45.   Yu, L.; Gong, B.; Duan, W.; Fan, C.; Zhang, J.; Li, Z.; Xue, X.; Xu, Y.; Meng, D.; Li, B.; et al. Melatonin
      ameliorates myocardial ischemia/reperfusion injury in type 1 diabetic rats by preserving mitochondrial
      function: Role of AMPK-PGC-1α-SIRT3 signaling. Sci. Rep. 2017, 7, 41337. [CrossRef] [PubMed]
46.   Wölfler, A.; Caluba, H.C.; Abuja, P.M.; Dohr, G.; Schauenstein, K.; Liebmann, P.M. Prooxidant activity of
      melatonin promotes fas-induced cell death in human leukemic Jurkat cells. FEBS Lett. 2001, 502, 127–131.
      [CrossRef]
47.   Albertini, M.C.; Radogna, F.; Accorsi, A.; Uguccioni, F.; Paternoster, L.; Cerella, C.; De Nicola, M.;
      D’Alessio, M.; Bergamaschi, A.; Magrini, A.; et al. Intracellular pro-oxidant activity of melatonin deprives
      U937 cells of reduced glutathione without affecting glutathione peroxidase activity. Ann. N. Y. Acad. Sci.
      2006, 1091, 10–16. [CrossRef] [PubMed]
48.   Cristofanon, S.; Uguccioni, F.; Cerella, C.; Radogna, F.; Dicato, M.; Ghibelli, L.; Diederich, M. Intracellular
      prooxidant activity of melatonin induces a survival pathway involving NF-kappaB activation. Ann. N. Y.
      Acad. Sci. 2009, 1171, 472–478. [CrossRef] [PubMed]
You can also read